Antimalarial, Antitrypanosomal, and Antileishmanial Activities and

Biologie et Evolution des Parasites, CNRS - EP1790,. 61 rue Buffon, 75005 Paris, France, and Tibotec, L11 Gen. de Wittelaan, B-32800 Mechelen, Bel...
0 downloads 0 Views 502KB Size
2646

J. Med. Chem. 2000, 43, 2646-2654

Articles Antimalarial, Antitrypanosomal, and Antileishmanial Activities and Cytotoxicity of Bis(9-amino-6-chloro-2-methoxyacridines): Influence of the Linker# Sophie Girault,† Philippe Grellier,‡ Amaya Berecibar,⊥,† Louis Maes,§ Elisabeth Mouray,‡ Pascal Lemie`re,† Marie-Ange Debreu,† Elisabeth Davioud-Charvet,† and Christian Sergheraert*,† UMR 8525 CNRS, Universite´ de Lille II, Institut de Biologie et Institut Pasteur de Lille, 1 rue du Professeur Calmette, BP 447, 59021 Lille, France, Muse´ um National d’Histoire Naturelle, Biologie et Evolution des Parasites, CNRS - EP1790, 61 rue Buffon, 75005 Paris, France, and Tibotec, L11 Gen. de Wittelaan, B-32800 Mechelen, Belgium Received November 3, 1999

Forty bis(9-amino-6-chloro-2-methoxyacridines), in which acridine moieties are joined by alkanediamines, polyamines, or polyamines substituted by a side chain, were synthesized and tested for their in vitro activity upon the erythrocytic stage of Plasmodium falciparum, trypomastigote stage of Trypanosoma brucei, and amastigote stage of Trypanosoma cruzi and Leishmania infantum as well as for their cytotoxic effects upon MRC-5 cells. Results clearly showed the importance of the nature of the linker and of its side chain for antiparasitic activity, cytotoxicity, and cellular localization. Among several compounds devoid of cytotoxic effects at 25 µM upon MRC-5 cells, one displayed IC50 values ranging from 8 to 18 nM against different P. falciparum strains while three others totally inhibited T. brucei at 1.56 µM. Introduction

Chart 1. Structure of Antimalarial Molecules

Malaria, trypanosomiasis, and leishmaniasis are major diseases in developing countries which continue to infect several hundreds of millions of people and which are responsible for a mortality rate in excess of 1 million per year. Present chemotherapies are proving inadequate, are toxic, or are becoming ineffective due to an increase in resistance. The trivalent arsenical drug melarsoprol is still widely used against the second stage of African trypanosomiasis in which Trypanosoma brucei rhodesiense or gambiense has invaded the central nervous system. The toxicity of this treatment is responsible for the death of approximately 5% of its recipients. Benznidazole, the only commercial drug still available for treatment during the chronic stage of Chagas’ disease, is no longer efficient, while its safety continues to be debated. Against Leishmania parasite, amphotericin B and pentamidine are toxic in therapeutic doses, while resistance to pentavalent antimonials is increasing. The effectiveness of chloroquine (CQ, Chart 1), the cheap, antimalarial mainstay for more than 50 years, is also being undermined by the evolution of resistant parasites.1,2 CQ is believed to exert its # Abbreviations: CQ, chloroquine; DAPI, 4,6-diamidino-2-phenylindole; MF, mefloquine; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (thiazolyl blue); PyBrop, bromotris(pyrrolidinophosphonium) hexafluorophosphate. * Corresponding author. Tel: (33) 3 20 87 12 11. Fax: (33) 3 20 87 12 33. E-mail: [email protected]. † Institut de Biologie et Institut Pasteur de Lille. ‡ CNRS - EP1790. § Tibotec. ⊥ Present address: Institut de Recherche Jouveinal, Parke-Davis, 3-9 rue de la Loge, 94265 Fresnes Cedex, France.

activity by inhibiting hemozoin formation in the digestive vacuole of the malaria parasite,3,4 though this has recently been questioned by Ginsburg and co-workers who have suggested that inhibition of the degradation of ferriprotoporphyrin IX by glutathione-dependent redox processes could be a second mode of action of CQ.5 Biochemical studies indicate that isolates of the CQresistant parasite accumulate less drug in the food vacuole than their more sensitive counterparts. However, opinion remains divided upon the mechanistic explanation for the reduction: (1) a rapid CQ efflux mechanism by CQ-resistant parasites,6 (2) an elevated pH in the food vacuole of the CQ-resistant parasites,7 (3) resistance being linked to a carrier-mediated CQ uptake,8 (4) resistance being linked to a reduced CQ affinity to ferriprotoporphyrin.9 CQ resistance may

10.1021/jm990946n CCC: $19.00 © 2000 American Chemical Society Published on Web 06/17/2000

Bis(9-amino-6-chloro-2-methoxyacridines)

Journal of Medicinal Chemistry, 2000, Vol. 43, No. 14 2647

involve several mechanisms; however its reversal by molecules such as verapamil, desipramine, and chlorpromazine suggests that an enhanced CQ efflux by a multidrug-resistant mechanism may be implicated.6,10 One possibility to overcome this mechanism is to design quinoline-based drugs which are not recognized by the proteins involved in drug efflux. In this regard, bulky bisquinolines were synthesized and suggested to be extruded with difficulty by a proteinaceous transporter.11 They were also discovered to inhibit the growth of CQ-sensitive and CQ-resistant parasites with similar efficiency though it was not proved that steric hindrance was the deciding factor for this.11-14 Further development of the most promising molecule Ro 47-773713 (Chart 1) as that of other bisquinolines, described earlier, piperaquine, hydroxypiperaquine, and dichloroquinazine,15 was suspended for reasons of toxicity. Acridine derivatives have been considered as potential antiprotozoal agents, e.g: 9-anilinoacridine topoisomerase II inhibitors show high levels of antileishmanial and antitrypanosomal activity.16 Furthermore, quinacrine, the 9-amino-6-chloro-2-methoxyacridine analogue of CQ (Chart 1), which was used clinically before CQ, is still an approved treatment of giardiasis. Sharing the same features as a weak diprotic base, it accumulates in the acidic food vacuole (pH ) 5) of Plasmodium and prevents hematin polymerization.4 Until now, bisacridines have been poorly studied for their antiparasitic activity, yet they, along with bisquinolines, may represent an alternative method to avoiding the efflux mechanism. In addition bisacridines offer the advantage of being viewed precisely by cellular fluorescence imaging. However, the presence of an acridine moiety in a molecule can lead to stronger interactions with DNA and generate cytotoxicity, especially in the case of bisintercalation. Bisintercalation requires a minimal distance between the acridine rings. Bis(9-amino-6chloro-2-methoxyacridines) corresponding to a spermine or spermidine linker were found to bisintercalate, while the removal of a single carbon (symmetrical linker -NH-(CH2)3-NH-(CH2)3-NH-) suppressed bisintercalation.17 It was also reported that acridine substituents play a role in intercalation and DNA recognition18,19 and that an increase of rigidity can favor binding affinity in relation to the subsequent decrease of the loss in entropy.20,21 Besides, for unsubstituted bisacridines, an evolution from monofunctional to bifunctional interaction was found by lengthening the methylene chain from four carbons to six.19,22,23 We have therefore undertaken the synthesis and an in vitro antiparasitic activity study upon Plasmodium falciparum of three series of bisacridines in which aromatic rings are joined by alkanediamines, polyamines,

Chart 2. Bisacridines of the A, B, and C Series

Scheme 1. Synthesis of Bisacridines 1-9a

a

Reagents: K2CO3, DMF.

or polyamines substituted by a side chain (Chart 2). In the latter case, the length of the linker proved unfavorable to bisintercalation, while the nature of the side chain could contribute to either a diminishing or canceling of the interaction with the phosphate groups of human DNA and consequently to a decrease of cytotoxicity. Antiparasitic efficacy of these compounds against Trypanosoma cruzi, Trypanosoma brucei, and Leishmania infantum, as well as the evaluation of their cytotoxicity, is also reported. Chemistry In preference to the generally adopted method, employing phenol, as described by Canellakis et al,24 bisacridines 1-9 (A and B series, Chart 2) were synthesized according to the method described for 4-aminoquinolines preparation,25 by reacting polyamines with 3 equiv of 6,9-dichloro-2-methoxyacridine in DMF at reflux, in the presence of potassium carbonate as an inorganic base (Scheme 1). Chromatography on silica gel columns was the selected method for purification. In the C series (Chart 2), the side chain was fixed by coupling various carboxylic groups with the central, secondary amino group of compounds 6-8, using PyBroP reagent.26,27 Thus, bisacridines 10-25 were synthesized, as shown in Scheme 2, by coupling bisacridines 6-8 with appropriate N-Boc- and/or N-Fmoc-amino acids. N-Boc-amino protecting groups of compounds 10 and 21 were removed by treatment with a 1:1 mixture of TFA/CH2Cl2 to give deprotected analogues 26 and 29, respectively (Scheme 2). N-Fmoc-amino or Fmoc-ester protecting groups of compounds 19, 20, and 25 were removed by treatment with a 20:80 piperidine/DMF mixture to give deprotected analogues 27, 28, and 30,

2648

Journal of Medicinal Chemistry, 2000, Vol. 43, No. 14

Girault et al.

Scheme 2. Synthesis of Bisacridines 10-31a

a

Reagents: (a) PyBrop, DIEA, DMF; (b) TFA/CH2Cl2 (1:1); (c) piperidine/DMF (20:80); (d) K2CO3, H2O, MeOH.

Scheme 3. Synthesis of Bisacridines 32-34a

a

Reagents: (a) succinic anhydride, pyridine; (b) Boc2O, NH4HCO3, pyridine, DMF; (c) morpholine, PyBrop, DIEA, DMF.

respectively (Scheme 2). Acetyl protection of compound 23 was removed by treatment with an aqueous solution of K2CO3 to give deprotected compound 31 (Scheme 2). Thick-layer chromatography was used for purification. Compound 32 was synthesized, as shown in Scheme 3, by reaction of bisacridine 6 with succinic anhydride, while its amide analogue 33 was prepared by treating compound 32 with ammonium hydrogenocarbonate. The morpholinoamide derivative 34 was synthesized by coupling the carboxylic acid 32 with morpholine using the coupling method described in Scheme 2. In the case of bisacridines 35-40, fixation of the amino side chain was accomplished in two steps: the reaction of secondary amino compound 6 with 3-chloropropionyl chloride, using 1-ethylpiperidine as a base, followed by substitution of the remaining chloro group by the appropriate amine (Scheme 4).27 As a tertiary amine 1-ethylpiperidine was preferred to pyridine which

was previously reported to lead to a nonconvertible pyridinium salt.28 Biological Results In Vitro Antimalarial Activity upon P. falciparum. Initially all of the compounds were tested for their antimalarial activity upon the CQ-resistant strain FcB1R (IC50 CQ ) 138.6 nM, IC50 MF ) 6.9 nM). In the A series (Table 1), increasing the length of the alkanediamine linker proved unfavorable toward activity (IC50 values increased from 16 to 176 nM from 6 to 12 carbons). Values in the B series (Table 1), were consistent regardless of the distance between the nitrogen atoms of the linker (IC50 around 60 nM), except for the piperazine derivative 9 which proved more efficient (IC50 ) 17 nM). In comparison with all other compounds of the A and B series, this derivative also showed a unique and contrasting behavior at the

Bis(9-amino-6-chloro-2-methoxyacridines)

Journal of Medicinal Chemistry, 2000, Vol. 43, No. 14 2649

Scheme 4. Synthesis of Bisacridines 35-40a

a

Reagents: 3-chloropropionyl chloride, 1-ethylpiperidine, THF, then R1R2NH, THF.

Table 1. In Vitro Sensitivity of P. falciparum FcB1R Strain to Bisacridines 1-9 (A and B Series) molecule

series

1 2 3 4 5 6 7 8 9

A A A A A B B B B

central amine

n

NH NH NH piperazine

4 6 8 10 12 3 2 3 3

n′

IC50 (nM)a

3 2 4 3

18 ( 7b 16 ( 10b 19 ( 10b 57 ( 30b 176 ( 96b 55 ( 9c 63 ( 10b 61 ( 3b 17 ( 10d

a IC 50 CQ ) 138.6 ( 8.4 nM; IC50 MF ) 6.9 ( 0.3 nM. number of experiments: bn ) 3; cn ) 5; dn ) 4.

b-d

n,

cellular localization level. In confocal microscopy, while

infected erythrocytes incubated with 10 µM quinacrine displayed a weak fluorescence associated with the parasite food vacuole and the erythrocytic and parasitic membranes (data not shown), a high fluorescence, associated exclusively with the parasite, was observed with 1 µM compound 9 (Figure 1A). No labeling of the food vacuole and of the normal or infected erythrocyte cytosols/membranes was recorded. Fluorescence was mainly concentrated in structures which colocalized with propidium iodide or DAPI labeling, suggesting a concentration of compound 9 in the parasite nuclei (data not shown). In murine muscle L-6 cells, which were used as a control, no nuclear labeling was observed; the fluorescence of compound 9 was only associated with

Figure 1. Labeling of P. falciparum-infected erythrocytes (A, C) and mouse L-6 cells (B, D) using compounds 9 (A, B) and 1 (C, D). Infected cultures of P. falciparum were incubated with 1 µM bisacridine derivative and then with 10 µg/mL nonexchangeable lipid DIL C18(3) (see Experimental Section). DIL C18(3) permitted the visualization of the membranes of the infected and noninfected erythrocytes (arrow). Bisacridine derivatives accumulated specifically in the intracellular parasites (arrowhead). Food vacuole (FV) was not labeled. For compound 9, a high fluorescence was associated with a structure which colocalized with the parasite nucleus (N). L-6 cells were only labeled with bisacridine derivatives. Bar scale is in µm. A and B are images of one section, C and D are 3D reconstructions of a sequential image collection.

2650

Journal of Medicinal Chemistry, 2000, Vol. 43, No. 14

Table 2. In Vitro Sensitivity of P. falciparum FcB1R Strain to Bisacridines 10-40 (C Series) Ra

IC50 (nM)b

CO-CH(NHBoc)-CH3 CO-CH(NHBoc)-CH3 CO-CH(NHBoc)-CH3 CO-CH(NHBoc)-CH3 (D enantiomer) CO-CH2-NHBoc CO-CH(NHBoc)-CH2-CH(CH3)2 CO-CH(NHBoc)-CH(CH3)2 CO-CH(NHFmoc)-CH3 CO-CH2-NH-CO-CH(NHBoc)-CH3 CO-CH(NHBoc)-(CH2)4-NHFmoc CO-CH(NHBoc)-CH2-NHFmoc CO-CH(NHBoc)-CH2-NHBoc CO-CH(NHBoc)-CH2OBn CO-CH(NHBoc)-CH2OAc CO-CH(NHBoc)-CH2-COOC6H11 CO-CH(NHBoc)-CH2-COOFmoc CO-CH(NH2)-CH3 CO-CH(NHBoc)-(CH2)4-NH2 CO-CH(NHBoc)-CH2-NH2 CO-CH(NH2)-CH2-NH2 CO-CH(NHBoc)-CH2-COOH CO-CH(NHBoc)-CH2OH CO-(CH2)2-COOH CO-(CH2)2-CONH2 CO-(CH2)2-CO-morpholine CO-(CH2)2-morpholine CO-(CH2)2-piperidine CO-(CH2)2-pyrrolidine CO-(CH2)2-(4-methylpiperazine) CO-(CH2)2-piperazine CO-(CH2)2-NH(CH3)2

31 ( 2c 109 ( 3d 330 ( 59d 33 ( 2d 92 ( 19e 27 ( 4d 25 ( 7d 232 ( 85d 440f 273 ( 75d 71 ( 3d 55 ( 3d 138f 63 ( 15d 31 ( 4c 230 ( 16d >500d >500d 307f >500d >500d 247 ( 92d 270f 40 ( 10e 436 ( 56d 26 ( 2e 67 ( 11d 91 ( 19d 62 ( 8d >500f 39 ( 5d

molecule n n′ 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40

3 2 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3

3 2 4 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3

a Ac, acetyl; Bn, benzyl; Boc, tert-butoxycarbonyl; Fmoc, 9-fluorenylmethyloxycarbonyl. b IC50 CQ ) 138.6 ( 8.4 nM; IC50 MF ) 6.9 ( 0.3 nM. c-f n, number of experiments: cn ) 5; dn ) 3; en ) 4; fn ) 2.

vesicles (Figure 1B), which co-localized with lysosomespecific probe labeling (data not shown). The other members of the A and B series displayed a specific parasite labeling similar to that for compound 9 (Figure 1C). However, a weaker fluorescence was associated with the parasite nucleus while fluorescence was mainly associated with the parasite cytoplasm. When compared with compound 9, the A and B series revealed a marked difference in L-6 cells localization, showing a diffuse labeling within the cell cytosol and no concentration in lysosome-like vesicles (Figure 1D). In the C series (Table 2), activity proved to be highly dependent upon the nature of the Boc- and/or Fmocprotected amino acid residue fixed to the central nitrogen atom of the linker and upon the distance between the terminal nitrogen atoms. A high level of activity (IC50 values about 30 nM) was observed for the alanine derivatives 10 and 13 (n ) n′ ) 3), for both enantiomers,

Girault et al.

while their analogues 11 (n ) n′ ) 2) and 12 (n ) 3, n′ ) 4) were less active (IC50 of 109 and 330 nM, respectively). N-Boc-amino acids with longer hydrophobic side chains, leucine (compound 15) and valine (compound 16), were the most active, while the polarity of the side chains proved consistently unfavorable since the protected derivatives 10 and 19-25 were more active than their corresponding deprotected counterparts 26 and 27-31. Also, the succinimide derivative 33 revealed a better activity than its carboxylic analogue 32. Addition of a second amino acid residue (compound 18) was unfavorable. Amino derivatives 35-40 displayed IC50 values between 26 and 91 nM, with the exception of the piperazine derivative 39 (IC50 > 500 nM) which via its terminal nitrogen atom could play the role of hydrogen-bond donor. In this series, the absence of a protonable amino group led to the less active morpholine compound 34. Five compounds among the most active upon the CQ-resistant strain FcB1R (compounds 9, 10, 13, 15, and 16) were subsequently evaluated for their efficiency in inhibiting the growth of seven strains showing different degrees of resistance to CQ or MF (Table 3). An inverse correlation was observed for compounds 10, 13, 15, and 16, as recently reported for bisquinolines by Vennerstrom and coworkers,29 the more the parasites appeared resistant to CQ, the more they seemed sensitive to the compounds under investigation. On the contrary, piperazine compound 9 displayed quite similar efficiency (IC50 values between 8 and 18 nM), whatever the CQ resistance of the strain. No such correlation was observed with the resistance to MF. In Vitro Antitrypanosomal Activity upon T. cruzi, T. brucei, and L. infantum. Compounds of the series A and B (Table 4), with the exception of the piperazine derivative 9 and to a lesser extent compounds 4 and 5, were found to be very cytotoxic upon human diploid embryonic lung cells (MRC-5 cells) and upon mouse primary peritoneal macrophages used in the T. cruzi and L. infantum tests as well as upon T. brucei which is known to be highly sensitive to toxic molecules. In confocal imaging of T. cruzi epimastigotes, compound 9 was found to be mainly associated with a structure which colocalized with kinetoplast DNA (Figure 2), while compound 1 displayed a more diffuse fluorescence associated with both the kinetoplast and the cytosol (data not shown). In the C series (Table 5), only five compounds (25, 29-32) were devoid of cytotoxicity toward MRC-5 cells and mouse macrophages. Four of these molecules possess a polar group (amine, alcohol, or carboxylic acid)

Table 3. Efficiency of Five of the Most Active Compounds against FcB1R, To Inhibit Growth of Parasites Expressing Different Degrees of Resistance to CQ IC50 (nM)a

P. falciparum strain

CQ

MF

9

10

13

15

16

3D7 F32a GP1 FCR3 FCM29 W2 K1

20 ( 4 22 ( 2 39 ( 2 102 ( 6 216 ( 25 219 ( 16 267 ( 42

40 ( 10 62 ( 6 17 ( 3 36 ( 15 8(3 30 ( 3 12 ( 1

13 ( 3 8(2 11 ( 5 18 ( 6 13 ( 6 12 ( 2 16 ( 5

22 ( 10 66 ( 5 35 ( 12 ndb 11 ( 4 10 ( 5 12 ( 2

57 ( 6 89 ( 11 66 ( 5 32 ( 8 11 ( 4 18 ( 8 23 ( 5

48 ( 6 52 ( 4 63 ( 6 17 ( 6 13 ( 5 15 ( 7 14 ( 7

49 ( 10 57 ( 2 83 ( 6 22 ( 8 16 ( 6 40 ( 11 36 ( 6

a Parasites were considered resistant to CQ for IC 50 > 100 nM. IC50 values are the mean ( standard deviation of three independent experiments. b nd, not determined.

Bis(9-amino-6-chloro-2-methoxyacridines)

Figure 2. Labeling of T. cruzi epimastigotes with compound 9: A, phase contrast; B, labeling with 1 µM compound 9. Fluorescence was mainly associated with a structure which colocalized with kinetoplast DNA (K). Bar ) 3 µm.

in the side chain linked to the spacer. They were highly active against T. brucei (IC50 < 1.56 µM or close to 1.56 µM for compound 29) and only displayed a marginal activity against P. falciparum, T. cruzi, and L. infantum. Compound 29 inhibited trypanothione reductase (TR) from T. cruzi (IC50 ) 20 µM), while IC50 values for the other compounds were equal or superior to 50 µM in the presence of 50 µM trypanothione disulfide (data not shown); this result was not really surprising since compound 29 displays the structural requirements for TR recognition, e.g. the presence of aromatic rings and of a protonable side chain.27,28 Discussion Bisquinolines were used to overcome CQ resistance since they were likely to be extruded with difficulty by a proteinaceous transporter.11 They were also discovered to inhibit the growth of CQ-sensitive and CQ-resistant parasites. However, further development was suspended for reasons of toxicity. Monoacridine derivatives were reported to display high levels of antileishmanial and antitrypanosomal activity16 while quinacrine, the acridine analogue of CQ, was in clinical use before CQ. Bisacridines could constitute an alternative to bisquinolines and avoid the efflux mechanism related to CQ resistance. Compared with bisquinolines, bisacridines are sterically more hindering, yet they have similar pKa values (pKa of acridine rings ) 6.5) which permit ionization and consequent accumulation in the food vacuole of Plasmodium. However their potential interaction with DNA is greater, especially in the case of bisintercalation which can generate cytotoxicity. Bisintercalation requires a minimal distance between the acridine rings, different if the “excluded site model”30,31 (two aromatic rings have to be separated by at least two base pairs) is observed: g10.2 Å or not (two aromatic rings on either side of a base pair): g6.8 Å. These minimal distances were compared by Le Pecq and coworkers, with the distances calculated for bis(9-amino6-chloro-2-methoxyacridines) bound with amino linkers.17 Compounds corresponding to a spermine or spermidine linker displayed distances respectively of 16.1 and 11.2 Å and were found to bisintercalate, while the removal of a single carbon (symmetical linker -NH-(CH2)3-NH-(CH2)3-NH-) reduced the distance to 9.9 Å and suppressed bisintercalation. The latter named linker was therefore introduced into the majority of compounds 10-40 and was substituted by a variety of side chains in order to decrease possible interaction with DNA. Bisacridines 1-9, differing by the length, the nature of the linker, and the absence of a side chain, were also prepared and studied in comparison.

Journal of Medicinal Chemistry, 2000, Vol. 43, No. 14 2651

For compounds 1-9, no clear relationship appears between potential bisintercalation and cytotoxic effects upon MRC-5 cells. Among the compounds unable to bisintercalate (compounds 1, 6, and 7), cytotoxicity of 1 upon MRC-5 cells cannot be related to a definite target since this compound shows a diffuse labeling in murine L-6 cell cytosol (Figure 1D) while that of 6 and 7 might be explained by the possible inhibition of the polyamine metabolism.32 With compounds capable of bisintercalation (2-5 and 8-9), surprisingly no localization in the nucleus of murine L-6 cells was observed by confocal analysis. As with compound 1, a diffuse labeling of the cytoplasm was observed for compounds 2-5 and 8, in contrast to mepacrine which accumulated in the nucleus (data not shown). Compounds 4 and 5 displayed the lowest cytotoxic effects upon MRC-5 cells, while an absence of cytotoxicity was noted for compound 9. This difference in cytotoxicity could reflect differences in their cellular distribution. In fact, introduction of a piperazine leads to the capture of compound 9 in lysosome-like vesicules of L-6 cells (Figure 1B). The high level of activity of compound 9 upon P. falciparum could result from its greater accumulation in the parasite nucleus (Figure 1A) and its bisintercalating properties. The difference in composition of plasmodial and mammalian DNA may contribute to such a sizable accumulation.33,34 However, cytoplasmic labeling in the parasite was also observed for compound 9, as well as for compounds 2-5 and 8, suggesting that these compounds might also interfere with cytoplasmic targets. Each of the compounds 1-8 shows a high level of activity upon T. brucei, yet none of which can be retained due to their toxicity found upon murine macrophages and of the known sensitivity of T. brucei to toxic compounds. Therapeutic indexes of compounds 4 and 5 proved to be too weak for potential drug use in large numbers of patients benefitting only from a minimal medical supervision. Thus from series A and B, compound 9 is the sole representative worthy of note as a trypanocidal lead and especially as an antimalarial lead whose interest is enhanced by its consistent efficiency whatever the CQ sensitivity of the strain tested (Table 3). With the exception of 11 and 12, compounds 10-40 are derived from compound 6 with their side chain bound by an amide bond. As expected, the nature of their respective side chains had a strong influence on both cytotoxicity and activity against parasites. When bisacridines 10-40 were compared with starting compound 6, a global decrease in cytotoxic effects upon MRC-5 cells was observed while activity upon macrophages remained high. Only compounds 25 and 29-32 cause no or low levels of cytotoxicity upon both cell lines, and from among them, four include the presence of a polar group (alcohol, amino, or carboxylic). The relative polarity of this group renders likely a decreased cellular availability that would be responsible for this low cytotoxicity and also for the weak activity against all parasites, with the exception of T. brucei. T. brucei differs from P. falciparum, T. cruzi, and L. infantum by an extracellular growth that confers differences in biochemical pathways, especially in polyamine metabolism.35 Trypanothione reductase from T. cruzi, which is inhibited by compound 29 (IC50 ) 20 µM), cannot be

2652

Journal of Medicinal Chemistry, 2000, Vol. 43, No. 14

Girault et al.

Table 4. In Vitro Cytotoxicity of Compounds 1-9 upon MRC-5 Cells and in Vitro Activities toward Trypomastigote Forms of T. brucei and Amastigote Forms of T. cruzi and L. infantum molecule

cytotoxicity (%)

concn (µM)

25

1 2 3 4 5 6 7 8 9 Melb Benzb Amp Bb

100 100 100 90 100 100 100 100 0

12.5

6.25

100 100 90 80 100 100 80 0 20 0 100 100 100 100 100 100 0 0 CC50 ) 12.5 µM CC50 > 50 µM CC50 ) 25 µM

T. cruzi (%)a

T. brucei (%) 3.13

12.5

100 20 20 0 0 100 100 100 0

100 100 100 100 100 100 100 100 100

6.25

3.13

100 100 100 99 100 100 100 100 100 100 100 100 100 100 100 100 90 80 IC50 ) 200 nM

L. infantum (%)a

1.56

12.5

6.25

3.13

1.56

12.5

6.25

3.13

1.56

100 90 100 100 70 100 100 100 60

T 70 T 99 90 T T T 0

T 40 98 70 40 T T T 0

T 0 90 40 0 T T T 0

90 0 70 0 0 60 T T 0

T 40 T T T T T T 0

T 0 95 90 80 T T T 0

T 0 90 70 40 T T T 0

T 0 70 20 0 40 T T 0

IC50 ) 3130 nM

IC50 ) 50 nM

a

T, toxic upon mouse primary peritoneal macrophages used in the test. b Mel, melarsoprol; Benz, benznidazole; Amp B, amphotericin B. These reference drugs are included in each primary in vitro screening batch. Table 5. In Vitro Cytotoxicity of Compounds 10-40 upon MRC-5 Cells and in Vitro Activities toward Trypomastigote Forms of T. brucei and Amastigote Forms of T. cruzi and L. infantum molecule

T. brucei (%)a

cytotoxicity (%)

T. cruzi (%)b

L. infantum (%)b

concn (µM)

25

12.5

6.25

3.13

12.5

6.25

3.13

1.56

12.5

6.25

3.13

1.56

12.5

6.25

3.13

1.56

10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40

100 100 100 100 100 55 60 100 100 100 68 70 100 100 100 0 100 100 39 0 0 0 0 100 60 100 78 76 80 15 54

100 0 100 84 100 49 59 100 71 90 60 50 100 90 100 0 100 100 37 0 0 0 0 100 20 100 71 65 74 0 40

100 0 0 84 20 47 54 100 41 80 57 48 100 40 100 0 40 40 0 0 0 0 0 100 0 100 57 39 57 0 39

0 0 0 81 0 42 43 100 31 20 55 43 100 20 40 0 20 0 0 0 0 0 0 100 0 80 42 37 40 0 30

100 100 100 nd 100 nd nd 100 nd 100 nd nd 100 100 100 100 100 100 nd nd 100 100 100 100 100 100 nd nd nd nd nd

100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 90 100 100 95 100 100 100 100 100 100 100 100

100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 60 100 100 80 100 100 100 100 100 100 99 100

100 99 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 100 99 20 100 100 60 100 100 100 100 100 100 95 100

T 60 90 T 80 T T T T T T T T 70 98 40 0 99 T 0 80 80 0 T 60 T T T T 90 T

90 40 70 T 40 T T T 40 80 90 80 80 40 70 0 0 90 T 0 0 20 0 T 0 T T T T 0 T

40 0 20 95 0 80 90 0 0 20 20 40 40 0 20 0 0 40 40 0 0 0 0 T 0 60 T T T 0 T

0 0 0 60 0 40 70 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 95 0 40 40 0 0 0 40

T 90 70 T 80 T T T T T T T T 90 T 20 0 40 T 0 0 40 0 T 0 T T T T T T

95 60 40 T 60 T T T T 40 95 99 90 80 80 0 0 0 T 0 0 0 0 T 0 T T T T 0 T

60 20 20 99 40 95 99 40 0 0 70 95 70 60 60 0 0 0 0 0 0 0 0 T 0 T T T T 0 T

40 0 0 95 0 90 95 0 0 0 20 80 40 0 20 0 0 0 0 0 0 0 0 T 0 T T T T 0 T

a

nd, not determined. b T, toxic upon mouse primary peritoneal macrophages used in the test.

retained as the target since this compound is totally inactive upon T. cruzi. Compounds 10, 13, and 15-16 display high levels of activity upon T. cruzi and L. infantum at 1.56 µM. They also show an intriguing inverse correlation toward P. falciparum growth inhibition for the seven strains studied and varying in their sensitivity to CQ. This might suggest either the presence of more sensitive target(s) in CQ-resistant parasites compared with CQ-sensitive ones or a higher accumulation inside the CQ-resistant parasites due to mechanism(s) which might be involved in CQ resistance. However, interest in these four compounds is rendered questionable by their cytotoxic effects upon MRC-5 cells and macrophages.

Conclusions From the hypothesis that bulky structures are extruded with difficulty by CQ-resistant strains of P. falciparum, a series of bisacridine derivatives (aliphatic di-, tri-, and tetramine) was prepared. A side chain with a variety of amino acid residues was attached to the polyamine linker, both to improve the weak solubility of this type of compound and to reduce the possible interaction with human DNA. The introduction of a piperazine moiety to the linker led to a unique behavior: a strong and selective activity upon Plasmodium, a localization outside of the food vacuole and associated mainly with the parasite nucleus along with a total absence of cytotoxic effects upon MRC-5 cells and

Bis(9-amino-6-chloro-2-methoxyacridines)

Journal of Medicinal Chemistry, 2000, Vol. 43, No. 14 2653

murine macrophages. Although no structure-activity relationship was evident in the C series with respect to bisintercalation, the presence of a polar group in the side chain attached to the linker afforded a reduction in antimalarial activity while favoring a specific activity upon T. brucei. Compounds 9, 25, 30, and 31, which satisfy both of the conditions required for antiparasitic drugs, e.g. safety and low cost, can be considered as good, new lead structures against P. falciparum and T. brucei.

the total parasite load (free trypomastigotes and intracellular amastigotes) compared with untreated control cultures. Scoring was performed microscopically. In Vitro Activity against Intracellular L. infantum Amastigotes. Primary mouse peritoneal macrophages were seeded in 16-well Lab-Tek culture slides at 30 000 cells/well. After 24 h, amastigotes of L. infantum (derived from the spleen of an infected donor animal) were added at an infection ratio of 10/1 together with 2-fold dilutions of the drug. The cultures were incubated at 37 °C in 5% CO2-95% air for 7 days. Treatment of uninfected control cultures was also included to determine a selectivity index. Drug activity was semiquantitatively scored as a percent (%) reduction of the total parasite load or the number of infected macrophages in Wright stained preparations. Scoring was performed microscopically. Confocal Microscopy. P. falciparum-infected red blood cells were maintained for 1 h under normal culture conditions, at 37 °C, in the presence of 1 µM bisacridine derivative. Following two washes (2000g, 5 min), cells were incubated in culture medium without serum for 15 min in the presence of 10 µg/mL DIL C18(3) (1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine perchlorate; Molecular Probes, Inc., Eugene, OR). After three further washes, cells were mounted on polyL-lysine-coated slides and immediately observed using a confocal laser scanning microscope (MRC 1024, Bio-Rad). In some experiments, following incubation with a bisacridine derivative, cells were fixed with 2% (v/v) formaldehyde in phosphatebuffered saline (PBS), washed twice, and incubated for 15 min with 20 µg/mL propidium iodide or 2 µg/mL DAPI (Sigma). Cells were observed after two further washes; no difference in labeling with bisacridine derivatives was noted between either living or fixed cells, or cells not incubated with DIL C18(3). For the labeling with mepacrine, infected erythrocytes were incubated with 10 µM drug for 1 h, washed twice, and immediately observed. Murine muscle L-6 cells were grown in 8-well Lab-Tek culture slides (Nunc, Inc., Naperville, IL) in DMEM supplemented with 5% (v/v) fetal calf serum, at 37 °C, in 5% CO2-95% air. Cells were incubated for 1 h in the presence of 1 µM bisacridine derivative and washed three times prior to observation. For double-labeling experiments using a lysosome-specific probe, cells were incubated simultaneously with a bisacridine derivative and LysoTracker Red DND-99 diluted according to the manufacturer’s recommendations (Molecular Probes). T. cruzi epimastigotes (Y strain) were maintained and grown in liver infusion medium, containing 10% (v/v) fetal calf serum, at 28 °C.40 Cells were washed twice in PBS and incubated with 1 µM bisacridine derivative in PBS, for 1 h. After three washes with PBS, cells were fixed with 1% (v/v) formaldehyde in PBS and incubated for 15 min with 2 µg/mL DAPI. Cells were observed following three further washes. Assays for TR Inhibition. Recombinant T. cruzi trypanothione reductase was produced from the SG5 Escherichia coli strain with the overproducing expression vector pIBITczTR. TR activity was measured at 21 °C in 0.02 M Hepes buffer, pH 7.25, containing 0.15 M KCl, 1 mM EDTA, and 0.2 mM NADPH with an enzyme concentration of 0.02 U mL-1. The reaction was promoted by the addition of the enzyme, and the subsequent NADPH oxidation was followed at 340 nm. IC50 of the different compounds was evaluated in the presence of 50 µM T(S)2 and 1% DMSO.

Experimental Section In Vitro P. falciparum Culture and Drug Assays. P. falciparum strains were maintained in continuous culture on human erythrocytes as described by Trager and Jensen.36 In vitro antiplasmodial activity was determined using a modification of the semiautomated microdilution technique of Desjardins et al.37 P. falciparum CQ-sensitive (3D7/unknown origin, F32a/Tanzania, GP1/Thailand), moderately CQ-resistant (FCR3/ Gambia), and CQ-resistant (FcB1R/Colombia, FCM29/Cameroun, K1/Thailand, W2/Indochina) strains were used in sensitivity testing. FcB1R, F32a, GP1, and FCM29 were strains obtained by limit dilution. Stock solutions of chloroquine diphosphate and test compounds were prepared in sterile distilled water and DMSO, respectively. Drug solutions were serially diluted with culture medium and added to asynchronous parasite cultures (0.5% parasitemia and 1% final hematocrite) in 96-well plates for 24 h, at 37 °C, prior to the addition of 0.5 µCi of [3H]hypoxanthine (1-5 Ci/mmol; Amersham, Les Ulis, France) per well, for 24 h. The growth inhibition for each drug concentration was determined by a relative comparison of the radioactivity incorporated in the treated culture with that in the control culture (without drug) maintained on the same plate. The concentration causing 50% inhibition (IC50) was obtained from the drug concentrationresponse curve, and the results are expressed as the mean determined from several independent experiments. The DMSO concentration never exceeded 0.1% and did not inhibit the parasite growth. Cytotoxicity Test upon MRC-5 Cells. A human diploid embryonic lung cell line (MRC-5, Bio-Whittaker 72211D) and primary peritoneal mouse macrophages were used to assess the cytotoxicity for host cells. The peritoneal macrophages were collected from the peritoneal cavity 48 h after stimulation with potato starch and seeded in 96-well microplates at 30 000 cells/ well. MRC-5 cells were seeded at 5 000 cells/well. After 24 h, the cells were washed and 2-fold dilutions of the drug were added in 200 µL of standard culture medium (RPMI + 5% FCS). The final DMSO concentration in the culture remained below 0.5%. The cultures were incubated with four concentrations of compounds (25, 12.5, 6.25, and 3.13 µM) at 37 °C in 5% CO2-95% air for 7 days. Untreated cultures were included as controls. For MRC-5 cells, the cytotoxicity was determined using the colorimetric MTT assay38 and scored as a percent (%) reduction of absorption at 540 nm of treated cultures versus untreated control cultures. For macrophages, scoring was performed microscopically. In Vitro Activity against T. brucei Trypomastigotes. Bloodstream forms of T. brucei were cultivated in HMI-9 medium.39 In a 96-well microplate, 10 000 hemoflagellates were incubated at different drug concentrations (12.5, 6.25, 3.13, and 1.56 µM) for 4 days. Parasite multiplication was measured colorimetrically (490 nm) following addition of MTT, which converts to an aqueous soluble, formazan product.38 In Vitro Activity against Intracellular T. cruzi Amastigotes. Primary mouse peritoneal macrophages were seeded in 96-well microplates at 30 000 cells/well. After 24 h, about 10 0000 trypomastigotes of T. cruzi were added per well together with 2-fold dilutions of the drug. The cultures were incubated at 37 °C in 5% CO2-95% air for 4 days. Following fixation in methanol and Giemsa staining, the drug activity was semiquantitatively scored as a percent (%) reduction of

Acknowledgment. We express our thanks to Ge´rard Montagne for NMR experiments, Vale´rie Landry for IC50 measurements upon TR, and Dr. Steve Brooks and Sandrine Delarue for proofreading. This work was supported by CNRS (GDR 1077, EP CNRS 1790, UMR CNRS 8525) and Universite´ de Lille II. Supporting Information Available: Details of chemical procedures and analytical data. This material is available free of charge via the Internet at http://pubs.acs.org.

2654

Journal of Medicinal Chemistry, 2000, Vol. 43, No. 14

References (1) Van Est, H. G.; Skamene, G. E.; Schurr, E. Chemotherapy of malaria: a battle against the odds? Clin. Invest. Med. 1993, 16, 285-293. (2) White, N. J. Antimalarial drug resistance: the pace quickens. J. Antimicrob. Chemother. 1992, 30, 571-585. (3) Dorn, A.; Stoffel, R.; Matile, H.; Bubendorf, A.; Ridley, R. G. Malarial haemozoin/β hematin supports haem polymerization in the absence of protein. Nature 1995, 374, 269-271. (4) Dorn, A.; Vippagunta, S. R.; Matile, H.; Jaquet, C.; Vennerstrom, J. L.; Ridley, R. G. An assessment of drug-haematin binding as a mechanism for inhibition of haematin polymerisation by quinoline antimalarials. Biochem. Pharmacol. 1998, 55, 727736. (5) Ginsburg, H.; Famin, O.; Zhang, J.; Krugliak, M. Inhibition of Glutathione-dependent Degradation of Heme by Chloroquine and Amodiaquine as a Possible Basis for Their Antimalarial Mode of Action. Biochem. Pharmacol. 1998, 56, 1305-1313. (6) Krogstad, D. J.; Gluzman, I. Y.; Kyle, D. E.; Oduola, A. M.; Martin, S. K.; Milhous, W. K.; Schlesinger, P. H. Efflux of chloroquine from Plasmodium falciparum: mechanism of chloroquine resistance. Science 1987, 238, 1283-1285. (7) Ginsburg, H.; Stein, W. D. Kinetic modeling of chloroquine uptake by malaria-infected erythrocytes. Biochem. Pharmacol. 1991, 41, 1463-1470. (8) Wu¨nsch, S.; Sanchez, C. P.; Gekle, M.; Grobe-Wortmann, L.; Wiesner, J.; Lanzer, M. Differential stimulation of the Na+/H+ exchanger determines chloroquine uptake in Plasmodium falciparum. J. Cell. Biol. 1998, 140, 335-345. (9) Bray, P. G.; Janneh, O.; Raynes, K. J.; Mungthin, M.; Ginsburg, H.; Ward, S. A. Cellular uptake of chloroquine is dependent on binding to ferriprotoporphyrin IX and is dependent on NHE activity in Plasmodium falciparum. J. Cell. Biol. 1999, 145, 363376. (10) Reed, M. B.; Saliba, K. J.; Caruana; S. R.; Kirk, K.; Cowman A. F. Pgh1 modulates sensitivity and resistance to multiple antimalarials in Plasmodium falciparum. Nature 2000, 403, 906909. (11) Vennerstrom, J. L.; Ellis, W. Y.; Ager, A. L., Jr.; Andersen, S. L.; Gerena, L.; Milhous, W. K. Bisquinolines. 1. N,N-Bis(7chloroquinolin-4-yl)alkanediamines with potential against cloroquine-resistant malaria. J. Med. Chem. 1992, 35, 2129-2134. (12) Raynes, K.; Galatis, D.; Cowman, A. F.; Tilley, L.; Deady, L. W. Synthesis and activity of some antimalarial Bisquinolines. J. Med. Chem. 1995, 38, 204-206. (13) Ridley, R. G.; Matile, H.; Jaquet, C.; Dorn, A.; Hofheinz, W.; Leupin, W.; Masciadri, R.; Theil, F.-P.; Richter, W. F.; Girometta, M.-A.; Guenzi, A.; Urwyler, H.; Gocke, E.; Potthast, J.-M.; Csato, M.; Thomas, A.; Peters, W. Antimalarial activity of the Bisquinoline trans-N1,N2-Bis(7-chloroquinolin-4-yl)cyclohexane1,2-diamine: comparison of two stereoisomers and detailed evaluation of the S,S enantiomer, Ro 47-7737. Antimicrob. Chemother. 1997, 41, 677-686. (14) Cowman, A. F.; Deady, L. W.; Deharo, E.; Desneves, J.; Tilley, L. Synthesis and activity of some antimalarial Bisquinolinemethanols. Aust. J. Chem. 1997, 50, 1091-1096. (15) Basco, L. K.; Ruggeri, C.; Le Bras, J. Mole´ cules antipaludiques; Masson: Paris, 1994; pp 115-120. (16) Gamage, S. A.; Figgitt, D. P.; Wojcik, S. J.; Ralph, R. K.; Ransijn, A.; Mauel, J.; Yardley, V.; Snowdon, D.; Croft, S. L.; Denny, W. A. Structure-activity relationships for the antileishmanial and antitrypanosomal activities of 1′-substituted 9-anilinoacridines. J. Med. Chem. 1997, 40, 2634-2642. (17) Le Pecq, J.-B.; Le Bret, M.; Barbet, J.; Roques, B. P. DNA polyintercalating drugs: DNA binding of Diacridine derivatives. Proc. Natl. Acad. Sci. U.S.A. 1975, 72, 2915-2919. (18) Gaugain, B.; Markovits, J.; Le Pecq, J.-B.; Roques, B.; Hydrogen Bonding in Deoxyribonucleic Acid Recognition. 1. Proton Nuclear Magnetic Resonance Studies of Dinucleotide-Acridine Alkylamide Complexes. Biochemistry 1981, 20, 3035-3042. (19) Chen, T. K.; Fico, R.; Canellakis, E. S. Diacridines, Bifunctional Intercalators. Chemistry and Antitumor Activity. J. Med. Chem. 1978, 21, 868-874. (20) Markovits, J.; Garbay-Jaureguiberry, C.; Roques, B. P.; Le Pecq, J.-B. Acridine dimers: influence of the intercalating ring and of the linking-chain nature on the equilibrium and kinetic DNAbinding parameters. Eur. J. Biochem. 1989, 180, 359-366.

Girault et al. (21) Denny, W. A.; Atwell, G. J.; Baguley, B. C.; Wakelin, L. P. G. Potential antitumor agents. Synthesis and antitumor activity of new classes of diacridines. The importance of linker chain rigidity for DNA binding kinetics and biological activity. J. Med. Chem. 1985, 28, 1568-1574. (22) Wakelin, L. P. G.; Romanos, M.; Chen, T. K.; Glaubiger, D.; Canellakis, E. S.; Waring, M. J. Structural limitations on the bifunctional interaction of Diacridines into DNA. Biochemistry 1978, 17, 5057-5063. (23) Wakelin, L. P. G.; Creasy, T. S.; Waring, M. J. Equilibrium constants for the binding of an homologous series of monofunctional and bifunctional intercalating diacridines to calf thymus DNA. FEBS Lett. 1979, 104, 261-265. (24) Canellakis, E. S.; Shaw, Y. H.; Hanners, W. E.; Schwartz, R. A. Diacridines: bifunctional intercalators. I. Chemistry, physical chemistry and growth inhibitor properties. Biochim. Biophys. Acta 1976, 418, 277-289. (25) Srivastava, S.; Tewari, S.; Chauhan, P. M. S.; Puri, S. K.; Bhaduri, A. P.; Pandey V. C. Synthesis of bisquinolines and their in vitro ability to produce methemoglobin in canin hemosylate. Bioorg. Med. Chem. Lett. 1999, 9, 653-658. (26) Coste, J.; Frerot, E.; Jouin, P. Coupling N-Methylated Amino Acids Using PyBroP and PyCloP Halogenophosphonium Salts: Mechanism and Fields of Application. J. Org. Chem. 1994, 59, 2437-2446. (27) Girault, S.; Davioud-Charvet, E.; Salmon, L.; Berecibar, A.; Debreu, M.-A.; Sergheraert, C. Structure-activity relationships in 2-aminodiphenylsulfides against trypanothione reductase from Trypanosoma cruzi. Bioorg. Med. Chem. Lett. 1998, 8, 1175-1180. (28) Girault, S.; Baillet, S.; Horvath, D.; Lucas, V.; Davioud-Charvet, E.; Tartar, A.; Sergheraert C. New potent inhibitors of trypanothione reductase from Trypanosoma cruzi in the 2-aminodiphenylsulfide series. Eur. J. Med. Chem. 1997, 32, 39-52. (29) Vennerstrom, J. L.; Ager, A. L., Jr.; Dorn, A.; Andersen, S. L.; Gerena, L.; Ridley, R. G.; Milhous, W. K. Bisquinolines. 2. Antimalarial N,N-Bis(7-chloroquinolin-4-yl)heteroalkanediamines. J. Med. Chem. 1998, 41, 4360-4364. (30) Crothers, D. M. Calculation of Binding Isotherms for Heterogeneous Polymers. Biopolymers 1968, 6, 575-581. (31) Bauer, W.; Vinograd, J. Interaction of Closed Circular DNA with Intercalative Dyes. II. The Free Energy of Superhelix Formation in SV40 DNA. J. Mol. Biol. 1970, 47, 419-435. (32) Saab, N. H.; West, E. E.; Bieszk, N. C.; Preuss, C. V.; Mank, A. R.; Casero, R. A., Jr.; Woster, P. M. Synthesis and Evaluation of Unsymmetrically Substituted Polyamine Analogues as Modulators of Human Spermidine/Spermine-N1-Acetyltransferase (SSAT) and as potential Antitumor Agents. J. Med. Chem. 1993, 36, 2998-3004. (33) Weber, J. L. A Review. Molecular Biology of Malaria Parasites. Exp. Parasitol. 1988, 66, 143-170. (34) Weber, J. L. Analysis of sequences from the extremely A + T-rich genome of Plasmodium falciparum. Gene 1987, 52, 103-109. (35) Wang, C. C. Validating targets for antiparasite chemotherapy. Parasitology 1997, 114, S31-S44. (36) Trager, W.; Jensen J. B. Human malarial parasites in continuous culture. Science 1976, 193, 673-677. (37) Desjardins, R. E.; Canfield, C. J.; Haynes, J. D.; Chulay, J. D. Quantitative assessment of antimalarial activity in vitro by a semiautomated microdilution technique. Antimicrob. Agents Chemother. 1979, 16, 710-718. (38) Mossman, T. Rapid Colorimetric Assay for Cellular Growth and Survival: Application to proliferation and Cytotoxicity Assays. J. Immunol. Methods 1983, 65, 55-63. (39) Hirumi, H.; Hirumi, K. Continuous cultivation of Trypanosoma brucei blood stream forms in a medium containing a low concentration of serum protein without feeder cell layers. J. Parasitol. 1989, 75, 985-989. (40) Camargo, E. P. Growth and differenciation in Trypanosoma cruzi: origin of metacyclic trypomastigotes in liquid media. Rev. Inst. Med. Trop. Sao Paulo 1964, 6, 63-100.

JM990946N